156
Views
10
CrossRef citations to date
0
Altmetric
Review

Six-month depot formulation of leuprorelin acetate in the treatment of prostate cancer

&
Pages 259-267 | Published online: 05 Jun 2009

Abstract

Hormonal deprivation therapy is well established for the treatment of locally advanced and metastatic prostate cancer, as well as the adjuvant treatment of some patients with localized disease. Long-acting gonadotropin releasing hormone (GnRH) agonists have become a mainstay of androgen deprivation therapy, due to their efficacy, tolerability, and convenience of use. One-month, 3-month, and 4-month depot leuprorelin formulations are well established and widely used to this end. Recently, a 6-month depot leuprorelin has been approved for use in advanced and metastatic prostate cancer patients. With similar efficacy and side effect profiles to earlier formulations, 6-month depot leuprorelin is a convenient treatment option for these patients. This review will highlight the role of GnRH agonists in the treatment of prostate cancer with a focus on the clinical efficacy, pharmacology, and patient-focused outcomes of the newer 6-month 45 mg depot leuprorelin formulation in comparison to available shorter-acting products.

Prostate cancer is one of the leading causes of cancer deaths among men in the United States. Current treatment options include radical prostatectomy, external beam radiotherapy, brachytherapy, and hormonal therapy. Hormonal therapy has become a mainstay of palliative treatment for patients with locally advanced or metastatic disease. Additionally, androgen deprivation is sometimes integrated with radiotherapy as definitive treatment of patients with localized disease. Selection factors for treatment include patient factors such as age, comorbidities, and patient preference; disease characteristics such as prostate-specific antigen (PSA), Gleason score, and stage; and psychosocial factors such as sexual function. Long-acting gonadotropin releasing hormone (GnRH) agonists have become widely accepted among patients and physicians as an alternative to earlier androgen-deprivation strategies, which included surgical castration and daily GnRH injections. Depot formulations of 1-month, 3-month, and 4-month dosages are well-established in the treatment of prostate cancer. This review will outline the role of hormonal deprivation therapy for prostate cancer patients, with an emphasis on the pharmacologic and clinical profile of a new 6-month depot formulation of leuprorelin acetate, also known as leuprolide acetate.

Therapeutic indications

Androgen suppression therapy is utilized as single-modality therapy for patients with localized disease as well as in conjunction with radiotherapy in patients with locally advanced disease or intermediate to high risk localized disease.

The rationale for using androgen deprivation with radiation therapy is based on the principle that cytoreduction through 2 modalities, namely hormones and radiation, may be more effective than local therapy alone. Movement toward this therapy began in patients with adverse tumor features such as bulky tumors, high PSA, and high Gleason score since they carried a poor prognosis with radiation therapy alone. Androgen suppression therapy is usually given in a neoadjuvant and concurrent manner, with additional adjuvant treatment following radiotherapy in those patients requiring longer-term treatment.

Neoadjuvant hormonal therapy could theoretically improve tumor control through 3 mechanisms: (1) Cytoreduction of tumor volume through apoptosis, (2) enhanced tumor cell kill because of radiation induced damage that leads to alternative pathways for apoptosis, or (3) improved radiosensitivity through reduced intra-tumoral hypoxia. While it is unclear which mechanism is most active, the cytoreductive mechanism is most strongly supported by in vitro and in vivo animal experiments and clinical investigations.Citation1

Multiple randomized-controlled trials have compared clinical outcomes of radiotherapy with adjuvant hormonal therapy to radiotherapy alone in prostate cancer patients with localized and locally advanced disease as well as patients with regional nodal involvement.Citation2Citation11 A meta-analysis of five consecutive Radiation Therapy Oncology Group (RTOG) phase III trials, including 2742 men treated between 1975 and 1992 showed improved outcomes in some groups of patients who received hormonal deprivation therapy. Patients were stratified into four prognostic risk groups based on Gleason score, clinical T-stage, and pelvic nodal involvement. PSA was not included because most patients were treated in the pre-PSA era. While low-risk patients (Gleason score 2–6 and T1–2Nx) did not benefit from adjuvant hormonal therapy, the intermediate- and high-risk groups (T3, N+, or Gleason score > 6) had improved overall survival and 8-year disease-specific survival with the addition of long-term hormonal therapy.Citation12Citation13 In the intermediate- and high-risk groups, 8-year overall survival improved from 45% to 61% and 28% to 44%, respectively, and 8-year disease-specific survival improved from 70% to 88% and 42% to 69%, respectively, when long-term hormonal therapy was used.

Many investigators have adopted a combined neoadjuvant-concurrent-adjuvant approach to hormonal therapy. D’Amico and colleagues conducted a prospective randomized trial in intermediate- and high-risk patients. Patients had localized disease but were required to have at least one adverse feature, defined as a PSA of greater than 10, a Gleason score of greater than 7, or radiographic evidence of extraprostatic disease on magnetic resonance imaging. Intermediate-risk patients were those with a Gleason score of 7 and PSA < 20 or with PSA 10–20 and Gleason ≥ 6. Patients were randomized to either radiation therapy alone to a dose of 70 Gy in 7 weeks to a localized prostate volume versus the same radiotherapy with 6 months of androgen suppression which was started 2 months before radiation and continued during radiation and then for 2 months after radiation. With a median follow-up of 4.5 years, the authors observed a significantly higher survival (88% vs 78% at 5 years), lower prostate cancer specific mortality (3.8% versus 0% at 5 years), and higher survival free of salvage androgen suppressive therapy (82% vs 57% at 5 years).Citation14

The timing and duration of androgen deprivation are still debatable topics, but many investigators believe that 6 months of androgen deprivation is appropriate for intermediate-risk cases while a longer duration (2 to 3 years) is more appropriate for patients with high-risk disease. This neoadjuvant-concurrent-adjuvant approach has also been adopted for longer-term androgen deprivation. Current studies in the RTOG typically employ 2 months of hormone treatment prior to RT and then continue for a total of 2 years in high risk patients. Ongoing studies are also examining whether shorter durations of hormonal therapy will suffice since androgen deprivation can confer significant toxicity.

Androgens and the prostate

Testicular hormone secretion has long been known to influence prostate growth. In the 18th century, surgical castration was noted to cause prostate atrophy in adult animals and halt prostate growth in younger animals.Citation15 In the late 19th century, castration was utilized to treat urinary retention caused by prostatic hyperplasia. In one early case series published in 1895, over half of the patients experienced improvement of urinary symptoms after surgical castration.Citation16

Androgen deprivation therapy (ADT) was first proposed as a treatment for prostate cancer in 1940 when castration was utilized to provide pain relief, stabilize tumor burden, and reduce serum acid phosphatase in prostate cancer patients with osseous metastatic tumors.Citation17,Citation18

Given the morbidity of surgical orchiectomy and associated clinical side effects, alternative anti-androgenic measures have been sought. One early approach attempted chemical castration through injection of the female hormones stilbestrol and hexestrol to neutralize the effect of testicular androgens.Citation18 The mechanism of this effect was discovered many years later: estrogen inhibits hypothalamic GnRH release through a negative feedback mechanism.Citation19

In subsequent decades, the role of the hypothalamic-pituitary axis in controlling testosterone production, namely through the secretion of GnRH, was elucidated. GnRH is secreted in a pulsatile fashion by the hypothalamus. This stimulates the anterior pituitary gland to release gonadotropin, which in turn acts on Leydig cells within the testes to stimulate testosterone production. Testosterone and adrenal androgens can be converted to dihydrotestosterone (DHT), a more potent hormone, within the cytoplasm of prostatic cells by the enzyme 5-alfa-reductase.Citation20 DHT binds to an androgen receptor within the cytosol, then translocates into the nucleus where it can affect gene synthesis, the production of proteins such as prostate-specific antigen (PSA), and cellular functions such as proliferation, growth, and cell death.

In subsequent years, multiple classes of anti-androgenic drugs have been developed to target the different steps of elucidated androgen regulatory pathways. These drug classes include GnRH agonists, GnRH antagonists, anti-androgens, 5-alfa reductase inhibitors, and the antifungal agent ketoconazole.Citation21Citation23 In clinical practice, synthetic GnRH agonists, especially in depot formulations, remain the mainstay of hormonal ablative therapy.

Currently available leuprorelin formulations

In 1971, GnRH was first isolated and characterized in the laboratory.Citation24 Leuprorelin, a synthetic analogue of GnRH, was first synthesized for clinical use in 1974.Citation25 Leuprorelin has a longer half-life and is 80 times more potent than naturally occurring GnRH, because of its enhanced binding affinity and increased resistance to degradation by peptidases.Citation26 Alterations in the chemical structure, including substitution of a D-amino acid for glycine at position 6 and deletion of glycine at position 10 with the insertion of ethylamide, are responsible for these properties.Citation27

The clinical benefits of GnRH analogues in prostate cancer patients were first described by Tolis et al in 1982. Ten patients with locally advanced or metastatic prostate cancer were treated with leuprorelin given as daily subcutaneous injections or twice daily intranasal applications for a period of 6 weeks to 12 months. During the treatment period, those patients with urinary obstruction noted improvement in urine flow, and those with osseous metastases reported decreased bone pain.Citation28

In 1985, leuprorelin was approved by the US Food and Drug Administration (FDA) for the palliative treatment of advanced prostate cancer.Citation29 Although the first clinical uses of synthetic GnRH required cumbersome daily injections, usually 1 mg given subcutaneously or intramuscularly, development of this medication revolutionized hormonal ablation therapy by allowing men to avoid the psychological and emotional consequences of surgical castration.

Technological advances have fostered the development of multiple long-acting depot formulations of GnRH in order to improve convenience of use, quality of life, and patient compliance. The first long-acting formulation was a monthly injection approved by the FDA in 1989 for treatment of advanced prostate cancer.Citation30 In the US, leuprorelin is now available in monthly (7.5 mg), 3-monthly (22.5 mg), 4-monthly (30 mg), and 6-monthly (45 mg) dosages.

The 3-monthly and 4-monthly formulations were approved by the FDA in 2002 and 2003, respectively, for treatment of advanced prostate cancer. They gained wide popularity, and within a year of its release, the 4-monthly formulation accounted for 40% of the market.Citation31 Most recently, the FDA approved a 45 mg 6-month depot leuprorelin in December 2004 for the palliative treatment of locally advanced or metastatic prostate cancer. These long-acting formulations are easy to use and require less effort on the part of both patient and clinician, which may in turn improve patient compliance, clinical efficacy, and outcomes. Additionally, unlike surgical castration, these medications are reversible, which can protect patients from the long-term consequences of a hypo-androgenic state such as osteopenia and muscle atrophy.

In Europe, a 3.75 mg 1-month depot leuprorelin formulation and an 11.25 mg 3-month depot formulation are available for prostate cancer treatment. In the US, these dosages are FDA approved only for the treatment of endometriosis, fibroids, and precocious puberty, but not for the treatment of prostate cancer. A 6-monthly 30 mg leuprorelin dosage has been developed and tested for efficacy and clinical safety in a recent European multicenter prospective trial.Citation32

Sustained release parenteral depot formulations administer hydrophilic leuprorelin that has been entrapped in biodegradable, highly lipophilic synthetic polymer microspheres. The preparation is made by dissolving both the drug and the biodegradable polymer in an organic solvent, with resultant in situ microsphere formation. Leuprorelin is released from the microspheres at a functionally constant rate over 1, 3, 4, or 6 months, depending on the polymer type.Citation33,Citation34 The delivery system for the six-month depot formulation utilizes a DL-lactide-co-glycolide polymer, with an 85:15 DL-lactide to glycolide molar ratio.

A 12-month subcutaneous implant was developed and briefly available for clinical use after approval by the FDA in 2000 for treatment of locally advanced or metastatic prostate cancer. However, the drug did not gain popularity and was eventually discontinued by the manufacturer in December 2007.Citation35,Citation36 There may have been less interest in this formulation compared to the depot injections because the 12-month implant entailed a surgical procedure for administration and required good follow-up with patient return for implant removal 12 months later.

Mode of action

Leuprorelin, is a synthetic nonapeptide analogue of naturally occurring GnRH. The chemical name is 5-oxo-L-prolyl-L-histidyl-L-tryptophyl-L-seryl-L-tyrosyl-D-leucyl-L-leucyl-L-arginyl-N-ethyl-L-prolinamide acetate (salt).Citation37

When given continuously, leuprorelin inhibits pituitary secretion of gonadotropin, which in turn suppresses testicular and ovarian steroidogenesis. Initial administration of leuprorelin causes an increase in gonadotropin levels, which can last for several weeks, leading to a rise in gonadal steroid production during that time. With continuous administration, there is eventual suppression of gonadotropin release within 2 to 4 weeks. In males, testosterone is reduced to a level below the castrate threshold, or ≤50 ng/dL. Upon removal of the drug, this effect is reversible.Citation33

Pharmacokinetics of leuprorelin depot

Because it is a peptide, leuprorelin is not active when given orally and is usually administered through a subcutaneous or intramuscular route.

Pharmacokinetic studies showed that mean peak plasma leuprorelin concentrations (Cmax) were 13.1, 21.8, 47.4, 54.5 and 53 μg/L after injection of 3.75, 7.5, 11.25, 15, and 30 mg depot formulations, respectively, and occurred within 1 to 3 hours of administration. After subcutaneous injection of 1 mg of a non-depot formulation, mean Cmax was 35 μg/L and occurred 36 to 60 minutes after injection.Citation39 Following injection of 45 mg 6-month depot formulation, there was an early rise in Cmax to 82.0 ng/mL 4.5 hours after the initial administration. On the second injection, 6 months after the initial treatment, mean Cmax was 102 ng/mL and occurred 4.5 hours after the second injection. After these initial increases, mean serum levels remained constant within the 0.2 to 2 ng/mL range.Citation37

The mean volume of distribution of leuprolide after bolus administration in a group of healthy male volunteers was 27 L.Citation39 After a single subcutaneous injection of 1 mg, 3.75 mg, 7.5 mg, or 15 mg leuprorelin, the mean volumes of distribution were 36 L, 33 L, and 27 L, respectively.

Total body clearance and elimination half-life were 9.1 L/hour and 3.6 hours, respectively, after a 1 mg subcutaneous administration. After intravenous bolus, these values were 8.3 L/hour and 2.9 hours, respectively.Citation33

To our knowledge, there is no published data documenting the volume of distribution, elimination half-life, or clearance of the 45 mg 6-month depot formulation of leuprorelin. The pharmacokinetics of leuprorelin have not been evaluated in a population of patients with compromised kidney or liver function.

Efficacy

Serum testosterone

The efficacy of the 6-month depot leuprorelin formulation was evaluated in a 12-month, open label, multicenter trial. One hundred eleven patients with prostate adenocarcinoma were enrolled. Inclusion criteria were stage > T1, WHO performance score 0–2, and life expectancy ≥ 1 year. Patients received leuprorelin 45 mg subcutaneously on days 1 and 168, a six month interval.Citation38 The primary endpoint of this study was a decrease in serum testosterone to a level equivalent to or below that resulting from surgical castration. Historically, the FDA had established the castrate threshold, or the testosterone level consistent with that obtained after surgical orchiectomy, to be 50 ng/dL.Citation41 However, this was largely based on the sensitivity of available laboratory assays at the time. With the development of newer assay techniques, substantially lower testosterone levels (15 ng/dL) have been observed in men after bilateral orchiectomy, which has led to reassessment of the historical threshold level by the medical community. The National Comprehensive Cancer Network amended its guidelines to suggest that serum testosterone level ≤ 20 ng/dL reflected optimal control of testosterone after surgical or chemical castration, and several other expert opinions have been published on this matter in agreement.Citation41,Citation42 In light of this, the 6-month depot leuprorelin efficacy study evaluated the number of patients with serum testosterone level below two separate thresholds: 50 ng/dL and also below 20 ng/dL, measured on at least two occasions at least 1 week apart. Serum PSA and gonadotropin levels and treatment-related toxicity were also assessed.

One hundred three of the 111 enrolled patients received both injections. There was an initial rise in testosterone level, which increased to a mean level of 588ng/dL by day 2. By day 28, 108 patients (97%) had achieved a serum testosterone level at or below the castrate threshold (50 ng/dL), and 92 (83%) had achieved optimal control of testosterone (≤20 ng/dL). After 12 months, 102 of the 103 (99%) patients who completed the study had testosterone levels below castrate threshold, and 91 patients (88%) had optimal control of testosterone. Median time to reach castrate level was 21 days.

One patient did not experience castrate level androgen suppression and was removed from the study at day 85. During a follow-up period of 12 months, only 1 patient experienced breakthrough testosterone levels above 50 ng/dL.Citation38

In comparison to conventional GnRH agonists, clinical studies indicate that 6-month depot leuprorelin may be more efficacious, although there are no prospective trials comparing these different formulations. Five percent to 17% of patients treated with daily GnRH injections do not reach the historical castrate level (≤50 ng/dL), and 13% to 34% fail to achieve optimal control of testosterone (≤20 ng/dL).Citation40,Citation43Citation47

Six-month depot leuprorelin appears to have similar efficacy to the other available depot formulations. The proportion of patients achieving optimal testosterone control (≤20ng/dL) after 6 to 8 months of treatment with 6-monthly (45 mg), 4-monthly (30 mg), 3-monthly (22.5 mg), and monthly (7.5 mg) formulations were 94%, 90%, 97.5%, and 94%, respectively.Citation48Citation50 Among the different formulations, 98% tp 100% of patients who completed the study had castrate level serum testosterone at study completion.

Transient testosterone escape (level ≥ 50 ng/dL on two separate occasions at least a week apart) was observed in no patients treated with the 7.5 mg monthly or 22.5 mg 3-monthly formulations. Three patients treated with the 30 mg 4-monthly formulation had transient testosterone breakthrough at 4 months, and 1 of these patients had a second breakthrough at 8 months. This patient had a small but clinically insignificant rise in PSA from 2.2 to 2.6 during the first breakthrough response but did not exhibit any other PSA elevations during treatment. One patient treated with the 45 mg 6-monthly formulation had transient testosterone breakthrough.Citation38,Citation48Citation50

Testosterone breakthrough is seen in about 5% of patients treated with conventional, daily leuprorelin injections.Citation40 There are several theories to explain this phenomenon, including increased GnRH receptor density during treatment, alternate GnRH receptor expression, phosphorylation of the GnRH receptor or its downstream G-protein, and uncoupling of the GnRH receptor and its target G-protein.Citation51

A recent European multicenter, prospective randomized trial compared treatment of prostate cancer patients over 12 months with an 11.25 mg 3-monthly formulation (currently approved for use in Europe), with two different 6-month depot formulations: a 22.5 mg dose and a 30 mg dose. One hundred seventy-eight patients with newly diagnosed or relapsed prostate cancer of any grade or stage were enrolled in the trial. Because of inferior response rates and efficacy of the 22.5 mg 6-month depot formulation, it was not selected for submission for approval in European countries, and therefore results from that arm were not published. The remaining two arms had similar efficacy and safety profile. After 12 months of treatment, 100% versus 98% of patients treated with 11.25 mg 3-month depot and 30 mg 6-month depot leuprorelin, respectively, had serum testosterone levels below castrate level (≤50 ng/dL), and 90% versus 81% had optimal testosterone control (≤20 ng/dL), respectively. These differences were not statistically significant.Citation38 As a result of this study, the 30 mg 6-month depot formulation has been submitted for approval for use in the treatment of prostate cancer patients in Europe.

Several recent studies indicated that depot leuprorelin formulations may be efficacious for longer than the recommended dosing intervals. Pathak et al conducted a prospective study in which 42 patients were treated with 22.5 mg subcutaneous injections of leuprorelin every 3 months, on day 1, after 12 weeks, and after 24 weeks. Serum testosterone levels were monitored at baseline, after 12 weeks, after 24 weeks, and monthly thereafter. If patients were still at castrate levels after 24 weeks, the subsequent injection was withheld until testosterone exceeded 50 ng/dL. After a median follow-up of 18 months, the median dosing interval was 6 months, with a range of 5 to 12 months.Citation52

A recent prospective trial by Greil et al evaluated this type of testosterone-based treatment approach in patients treated with the 30 mg 4-month depot leuprorelin formulation. Serum testosterone levels were obtained at baseline and then monthly beginning 4 months after the first injection and 2 months after subsequent injections for a total of 18 months. The median number of days from injection to the first serum testosterone level ≥ 50 ng/dL was 159, 189, and 163 days for the first, second, and third treatment cycle, respectively.Citation53

A multicenter randomized controlled trial by Gulley et al assessed time to testosterone recovery in 159 patients treated with two 6-month cycles of GnRH agonist therapy with two 3-month injections of leuprorelin (22.5 mg) or goserelin, another GnRH agonist.Citation54 Serum testosterone, DHT, and PSA were measured monthly until serum PSA progressed to a level above 5 ng/mL, at which point a second cycle was administered. Median time to testosterone normalization was 15.4 weeks and 18.3 weeks after cycles 1 and 2 respectively. Median time to DHT normalization was 15.2 weeks and 18.7 weeks after cycles 1 and 2, respectively.

These three studies suggest that patients treated in a testosterone-based manner can achieve sustained efficacy with exposure to fewer injections and lower drug levels, which may improve cost effectiveness and side effect profiles of GnRH agonist therapy. Additionally, periodic monitoring of serum testosterone levels is an important step in identifying patients who fail to achieve castrate levels or have breakthrough rises in testosterone while undergoing androgen deprivation treatment. For these reasons, some physicians are proponents of an individualized approach to hormonal deprivation therapy based upon patient serum testosterone levels as opposed to simply adhering to recommended dosing intervals.

Prostate-specific antigen

In patients treated with 45 mg 6-month depot leuprorelin, the percentage of patients with serum PSA levels within the normal range (<4 ng/mL) at baseline and after treatment was 25% and 96%, respectively. Mean PSA at baseline and after 12 months of treatment was 39.8 ng/mL and 1.2 ng/mL, respectively. This is similar to levels seen in patients treated with 7.5 mg monthly, 22.5 mg 3-monthly, and 30 mg 4-monthly dosages (Table ).Citation38,Citation48Citation50

Table 1 Effect of depot leuprorelin on serum prostate-specific antigen (PSA) levelCitation38,Citation48Citation50

Similar PSA levels were seen in the European randomized-controlled trial evaluating the lower dose depot formulations, including 11.25 mg 3-month depot and 30 mg 6-month depot leuprorelin. During months one through 12 of the study, PSA levels ranged from 0.2 to 1.0 ng/mL in the 3-monthly group and from 0.3 to 1.1 ng/dL in the 6-monthly group.

Serum gonadotropin

In the European multicenter clinical efficacy trial for 45 mg 6-monthly leuprorelin, there was an initial rise in gonadotropin as a result of leuprorelin’s GnRH agonist properties. Eight hours after injection with 45 mg depot leuprorelin, gonadotropin had increased to a mean of 37.9 mIU/mL. By day 7, mean gonadotropin decreased below baseline (6.9 mIU/mL), and it consistently declined over the first 19 weeks to a mean level of 0.1 mIU/mL. After the second injection, there was a transient, small increase in serum gonadotropin level to 0.2 mIU/mL on day 169, and gonadotropin levels remained steady at this level for the remainder of the study.Citation38

A similar pattern of gonadotropin surge was seen with administration of 7.5 mg monthly, 22.5 mg 3-monthly, and 30 mg 4-monthly dosages in the respective efficacy trials. Peak gonadotropin levels occurred on days 1 or 2 after leuprorelin administration, and decreased to below baseline between days 10 and 14.

An initial rise in testosterone occurs in parallel with this gonadotropin surge. Mean testosterone increased by 225 ng/dL by day 2, to 588 ng/dL, after the first injection of 6-month depot leuprorelin. A similar effect was seen with the other depot formulations.Citation55,Citation56 No clinically significant flare reactions in response to the early testosterone rise have been reported.Citation38,Citation48Citation50

Safety and tolerability

The majority of patients undergoing treatment with depot leuprorelin experience mild side effects. Fewer patients experience moderate adverse reactions, and severe toxicity is rarely reported. The most common side effects of leuprorelin are hot flashes, injection site reactions, fatigue, testicular atrophy, and gynecomastia.

After treatment with 45 mg 6-month depot leuprorelin, 82 (74%) of 111 participants reported 211 treatment-related side effects. One event was reported as severe, although the type of adverse reaction was not documented, and the other 210 events were mild to moderate.Citation38

Depot leuprorelin 45 mg 6-month has a similar side effect profile to the other depot formulations. Fifty-seven percent of patients treated with 22.5 mg 3-month depot leuprorelin experienced mild side effects, 12% experienced moderate side effects, and no patients experienced severe side effects.Citation49 Eighty-five percent of patients treated with 30 mg 4-month depot leuprorelin experienced treatment-related side effects, with 97% of these reactions being mild to moderate and 3% documented as severe hot flashes.Citation50 Seventy-four percent of patients treated with monthly depot leuprorelin experienced side effects of treatment. Most events were graded as mild to moderate; however 4% were considered severe, including hot flashes in 1 case and injection site burning in 4 instances.Citation48

Overall, with the exception of a notably higher rate of mild injection site reactions documented in patients receiving the 22.5 mg dosage, there were no substantial differences between the side effect profile of patients treated with the different depot formulations (Table ).

Table 2 Treatment-related adverse events of 6-month (45 mg), 4-month (30 mg), 3-month (22.5) mg, and 1-month (7.5 mg) depot leuprorelinCitation38,Citation48Citation50

No patients stopped treatment with the 45 mg, 22.5 mg, or 7.5 mg dosages due to treatment-related side effects. However, 3% of participants in the 4-month 30 mg depot leuprorelin clinical efficacy trial stopped because of side effects of treatment, although the specific reactions that caused patients to withdraw from the study were not documented.Citation50

Treatment compliance of patients enrolled in the above clinical efficacy trials was good. Of the patients enrolled for treatment with 7.5 mg monthly, 22.5 mg 3-monthly, 30 mg 4-monthly, and 45 mg 6-monthly leuprorelin, 98%, 98%, 91%, and 93% completed the 1-year treatment course, respectively.

In the European randomized controlled trial evaluating the lower-dose formulations (11.25 mg 3-month depot and 30 mg 6-month depot leuprorelin), 4% of patients withdrew from the study because of treatment-related adverse events. The most common side effects were hot flashes and injection-site reactions. Hot flashes occurred in 43% versus 34% of patients treated with 3-monthly and 6-monthly injections, respectively, and injection site reactions occurred in 2% and 11% of patients, respectively. About two-thirds of the injection site reactions were considered severe.Citation32

In summary, the available depot leuprorelin formulations are convenient and well tolerated with acceptable side effect profiles. Severe adverse events are rare, and patient compliance within published clinical studies is good. Few patients withdrew from the trials because of treatment-related side effects, and over 90% of patients completed the treatment course.

Conclusions

Hormonal deprivation therapy has become the mainstay of treatment for locally advanced and metastatic prostate cancer, as well as for the adjuvant treatment of patients with intermediate-risk or high-risk localized prostate cancer. Androgen deprivation has been shown to improve quality of life and prolong life in many patients who fall within these categories.

Surgical castration was the earliest form of androgen deprivation, but this has been replaced by chemical agents, which potentially have less physical and emotional impact than the surgical alternative. GnRH agonists are potent agents that block testosterone secretion from the testes, which encompasses 90% of the body’s testosterone production.Citation31 While the first synthetic GnRH analogues required daily injections, the introduction of long-acting synthetic GnRH agonists in the 1980s and 1990s revolutionized the hormonal treatment of prostate cancer. With their ease of use, tolerable side effect profile, and good efficacy, the depot formulations have gained wide acceptance from both patients and the medical community alike.

Today, 3-month and 4-month depot leuprorelin formulations are the most commonly used hormonal agents for the treatment of prostate cancer. Treatment with the shorter-acting variations such as 1-month depot and daily formulations presents more opportunities for patients to delay or altogether miss treatments, which can result in testosterone breakthrough and potentially deleterious effects on tumor control and symptom progression. The longer-acting formulations offer clinical benefit on these fronts by limiting the number of treatments involved in a therapeutic course.

The most recent addition to the hormonal deprivation armament is the 45 mg 6-month depot leuprorelin formulation. Further reducing the number of injections patients receive presents a number of advantages. First, treatment compliance will likely be improved with a decreased number of therapeutic delays or misses resulting in testosterone breakthrough. Second, patients may have fewer clinic visits, which are often anxiety-ridden and disruptive to their daily routine. Finally, since burning at the injection site is one of the most commonly reported treatment-related adverse events, the longer-acting formulations may improve the overall tolerability of the treatment by exposing patients to fewer injections.

In the US, multiple long-acting depot products (7.5 mg monthly, 22.5 mg 3-monthly, 30 mg 4-monthly, and 45 mg 6-monthly formulations) have been approved for use in prostate cancer. In Europe, several additional, lower-dose depot products have been approved for treatment of prostate cancer patients, including 3.75 mg monthly and 11.25 mg 3-monthly formulations.

Two clinical trials have shown that testosterone is often suppressed for longer than the recommended interval for a given depot product.Citation52,Citation53 Therefore, some physicians are proponents of an individualized, testosterone-based treatment system that utilizes periodic evaluation of serum testosterone levels to guide injection intervals and to detect non-responders and testosterone breakthrough.

In clinical trials thus far, 45 mg 6-month depot leuprorelin has similar clinical efficacy and tolerability, with acceptable rates of mild side effects and low rates of moderate to severe adverse events, compared with preceding shorter-acting depot formulations. The associated benefits of improved convenience, compliance, and tolerability will likely make this formulation popular among physicians and patients alike within the coming years.

Disclosures

The authors declare no conflicts of interest.

References

  • ZietmanALThe effect of androgen deprivation and radiation therapy on an androgensensitive murine tumor: an in vitro and in vivo studyCancer J Sci Am1997331369072305
  • BollaMLong-term results with immediate androgen suppression and external irradiation in patients with locally advanced prostate cancer (an EORTC study): A phase III randomised trialLancet200236010310612126818
  • BollaMImproved survival in patients with locally advanced prostate cancer treated with radiotherapy and goserelinN Engl J Med19973372953009233866
  • D’AmicoAV6-month androgen suppression plus radiation therapy vs radiation therapy alone for patients with clinically localized prostate cancer: A randomized controlled trialJAMA200429282182715315996
  • DenhamJWShort-term androgen deprivation and radiotherapy for locally advanced prostate cancer: Results from the Trans-Tasman Radiation Oncology Grou 96–01 randomised controlled trialLancet Oncol2005684185016257791
  • GranforsTCombined orchiectomy and external radiotherapy versus radiotherapy alone for nonmetastatic prostate cancer with or without pelvic lymph node involvement: A prospective randomized studyJ Urol1998159203020349598512
  • LaverdiereJThe efficacy and sequencing of a short course of androgen suppression on freedom from biochemical failure when administered with radiation therapy for T2–T3 prostate cancerJ Urol20041711137114014767287
  • PilepichMVPhase III trial of androgen suppression using goserelin in unfavorable-prognosis carcinoma of the prostate treated with definitive radiotherapy: Report of Radiation Therapy Oncology Group protocol 85–31J Clin Oncol199715101310219060541
  • PilepichMVAndrogen deprivation with radiation therapy compared with radiation therapy alone for locally advanced prostatic carcinoma: A randomized comparative trial of the Radiation Therapy Oncology GroupUrology1995456166237716842
  • PilepichMVPhase III radiation therapy oncology group (RTOG) trial 86–10 of androgen deprivation adjuvant to definitive radiotherapy in locally advanced carcinoma of the prostateInt J Radiat Oncol Biol Phys2001501243125211483335
  • PilepichMVAndrogen suppression adjuvant to definitive radiotherapy in prostate carcinoma – long-term results of phase III RTOG 85–31Int J Radiat Oncol Biol Phys2005611285129015817329
  • RoachM3rdFour prognostic groups predict long-term survival from prostate cancer following radiotherapy alone on Radiation Therapy Oncology Group clinical trialsInt J Radiat Oncol Biol Phys20004760961510837943
  • RoachM3rdPredicting long-term survival, and the need for hormonal therapy: A meta-analysis of RTOG prostate cancer trialsInt J Radiat Oncol Biol Phys20004761762710837944
  • D’AmicoAV6-month androgen suppression plus radiation therapy vs radiation therapy alone for patients with clinically localized prostate cancer: a randomized controlled trialJAMA20042927821715315996
  • AndroustosGJohn Hunter (1728–1793): fondateur de la chirugie scientifique etpercurseur de l’urologicProg Urol19988108710969894276
  • WhiteJWThe results of double castration in hyper-trophy of the prostateAnn Surg18952213
  • HugginsCStudies on prostate cancer: the effect of castration, of estrogen, and of androgen injection on serum phosphatases in metastatic carcinoma of the prostateCancer Res19401293
  • HugginsCStudies in prostatic cancerArch Surg194143209223
  • BrawerMKThe evolution of hormonal therapy for prostatic carcinomaRev Urol20013S11116986002
  • SandowJPharmacological studies on androgen suppression in therapy of prostate carcinomaAm J Clin Oncol198811S6S103291602
  • DebruyneFMJGonadotropin-releasing hormone antagonist in the management of prostate cancerRev Urol20046S253216985933
  • LoweFCIndications for use of ketoconazole in management of metastatic prostate canerUrology1990365415452247927
  • IversenPNonsteroidal antiandrogens: a therapeutic option for patients with advanced prostate cancer who wish to retain sexual interest and functionBJU Int200187475611121992
  • SchallyAVIsolation and properties of the FSH and LH-releasing hormoneBiochem Biophys Res Commun19714339394930860
  • FujinoMSynthetic analogs of lutenizing hormone releasing hormone (LH-RH) substituted in position 6 and 10Biochem Biophys Res Commun1974604064134608231
  • ChrispPLeuprorelin: a review of its pharmacology and therapeutic use in prostatic disordersDrugs Aging199114875091794035
  • MonahanMWSynthetic analogues of the hypothalamic lutenizing hormone releasing factor with increased agonist or antagonist propertiesBiochemistry197312461646204589943
  • TolisGTumor growth inhibition in patients with prostatic carcinoma treated with luteinizing hormone-releasing hormone agonistsProc Natl Acad Sci U S A198279165816626461861
  • SantenRJEvaluation of synthetic agonist analogue of gonadotropin-releasing hormone (leuprolide) on testicular androgen production in patients with carcinoma of prostateUrology1985252 Suppl5357
  • AkazaHLong-term clinical study on luteinising hormone-releasing hormone agonist depot formulation in the treatment of stage D prostatic cancer. The TAP-144-SR Study GroupJpn J Clin Oncol1992221771841518166
  • AbouelfadelZLeuprorelin depot injection: patient considerations in the management of prostatic cancerTher Clin Risk Manag2008451352618728847
  • TunnUWSafety and clinical efficacy of a new 6-month depot formulation of leuprorelin acetate in patients with prostate cancer in EuropeProstate Cancer Prostatic Dis200912838719030021
  • PeritiPClinical pharmacokinetics of depot leuprorelinClin Pharmacokinet20024148550412083977
  • SharifiRTherapeutic effects of leuprorelin microspheres in prostate cancerAdv Drug Deliv Rev19972812113610837568
  • FowlerJEEvaluation of an implant that delivers leuprolide for 1 year for the palliative treatment of prostate cancerUrology20005563964210792069
  • FowlerJESafety and efficacy of an implantable leuprolide delivery system in patients with advanced prostate cancerJ Urol200016473073410953135
  • Eligard.com [homepage on the Internet]Bridgewater, New Jersey [updated 2008 February; cited 2009 April 15]. Available from: www.eligard.com
  • CrawfordEDA 12-month clinical study of LA-2585 (45.0 mg): a new 6-month subcutaneous delivery system for leuprolide acetate for the treatment of prostate cancerJ Urol200617553353616406989
  • SennelloLTSingle-dose pharmacokinetics of leuprolide in humans following intravenous and subcutaneous administrationJ Pharm Sci1986751581603083092
  • OefeleinMGFailure to achieve castrate levels of testosterone during lutenizing hormone releasing hormone agonist therapy: the case for monitoring serum testosterone and a treatment decision algorithmJ Urol200016472672910953134
  • MillikanRUpdate of the NCCN guidelines for the treatment of prostate cancerOncology1997111801939430189
  • ZlottaARExpert opinion on optimal testosterone control in prostate cancerEur Urol Suppl200543741
  • McLeodDA phase 3 multicenter open-label randomized trial of abarelix versus leuprolide acetate in men with prostsate cancerUrology20015875676111711355
  • FontanaD3-month formulation of goserelin acetate (‘Zoladex’ 10.8-mg depot) in advanced prostate cancer: results from an Italian, open, multicenter trialUrol Int20037031632012740498
  • SardosyMFEndocrine effects, efficacy and tolerability of a 10.8-mg depot formulation of goserelin acetate administered every 13 weeks to patients with advanced prostate cancerBJU Int19998380180610368200
  • SharifiRLeuprolide Study GroupSerum testosterone suppression and potential for agonistic stimulation during chronic treatment with monthly and 3-month depot formulations of depot leuprolide acetate for advanced prostate cancerJ Urol20021681001100412187208
  • KhanMSAn evaluation of pharmacokinetics and pharmacodynamics of leuprorelin acetate 3M-depot in patients with advanced and metastatic carcinoma of the prostateUrol Int19986033409519419
  • Perez-MarrenoRA six-month, open-label study assessing a new formulation of leuprolide 7.5 mg for suppression of testosterone in patients with prostate cancerClin Ther2002241902191412501882
  • ChuFMA clinical study of 22.5 mg La-2550: a new subcutaneous depot delivery system for leuprolide aceate for the treatment of prostate cancerJ Urol20021681199120312187267
  • SartorOAn eight-month clinical study of LA-2575 30.0 mg: a new 4-month, subcutaneous delivery system for leuprolide acetate in the treatment of prostate cancerUrology2003623192312893343
  • RavivarapuHBParameters affecting the efficacy of a sustained release polymeric implant of leuprolideInt J Pharmacol2000194181191
  • PathakASDetermining dosing intervals for luteinizing hormone releasing hormone agonists based on serum testosterone levels: a prospective studyJ Urol20071772132213517509298
  • GreilSEfficacy over time of LHRH analogs in the treatment of PCa-a prospective analysis using serum testosterone to determine dosing intervalsUrology20097363163419110301
  • GulleyJLKinetics of serum androgen normalization and factors associated with testosterone reserve after limited androgen deprivation therapy for nonmetastatic prostate cancerJ Urol20081801432143718710748
  • SharifiRLeuprolide acetate (30-mg depot every four months) in the treatment of advanced prostate cancerUrology1998512712769495710
  • TunnUWComparison of LH-RH analogue 1-month and 3-month depot by their hormone levels and pharmacokinetic profile in patients with advanced prostate cancerUrol Int Suppl199860916